1
|
D’Ursi P, Rondina A, Zani A, Uggeri M, Messali S, Caruso A, Caccuri F. Molecular Mechanisms Involved in the B Cell Growth and Clonogenic Activity of HIV-1 Matrix Protein p17 Variants. Viruses 2024; 16:1048. [PMID: 39066211 PMCID: PMC11281387 DOI: 10.3390/v16071048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The human immunodeficiency virus (HIV-1) matrix protein p17 (p17) is released from infected cells as a protein capable of deregulating the biological activity of different cells. P17 variants (vp17s), more frequently detected in the plasma of HIV-1+ patients with rather than without lymphoma and characterized by amino acids insertions in their C-terminal region, were found to trigger B cell growth and clonogenicity. Vp17s endowed with B-cell-growth-promoting activity are drastically destabilized, whereas, in a properly folded state, reference p17 (refp17) does not exert any biological activity on B cell growth and clonogenicity. However, misfolding of refp17 is necessary to expose a masked functional epitope, interacting with the protease-activated receptor 1 (PAR-1), endowed with B cell clonogenicity. Indeed, it is worth noting that changes in the secondary structure can strongly impact the function of a protein. Here, we performed computational studies to show that the gain of function of vp17s is linked to dramatic conformational changes due to structural modification in the secondary-structure elements and in the rearrangement of the hydrogen bond (H-bond) network. In particular, all clonogenic vp17s showed the disengagement of two critical residues, namely Trp16 and Tyr29, from their hydrophobic core. Biological data showed that the mutation of Trp16 and Tyr29 to Ala in the refp17 backbone, alone or in combination, resulted in a protein endowed with B cell clonogenic activity. These data show the pivotal role of the hydrophobic component in maintaining refp17 stability and identify a novel potential therapeutic target to counteract vp17-driven lymphomagenesis in HIV-1+ patients.
Collapse
Affiliation(s)
- Pasqualina D’Ursi
- Institute of Technologies in Biomedicine, National Research Council, 20090 Segrate, Italy
| | - Alessandro Rondina
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
| | - Matteo Uggeri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
- Lifescience Innovation Good Healthcare Technology—LIGHT s.c.ar.l., 25123 Brescia, Italy
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy (M.U.)
- Centre for Advanced Medical and Pharmaceutical Research, “George Emil Palade” University of Medicine, Pharmacy, Science and Technology, 540142 Targu Mures, Romania
| |
Collapse
|
2
|
Zani A, Messali S, Bugatti A, Uggeri M, Rondina A, Sclavi L, Caccuri F, Caruso A. Molecular mechanisms behind the generation of pro-oncogenic HIV-1 matrix protein p17 variants. J Gen Virol 2024; 105. [PMID: 38687324 DOI: 10.1099/jgv.0.001982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024] Open
Abstract
HIV-1 matrix protein p17 variants (vp17s), characterized by amino acid insertions at the COOH-terminal region of the viral protein, have been recently identified and studied for their biological activity. Different from their wild-type counterpart (refp17), vp17s display a potent B cell growth and clonogenic activity. Recent data have highlighted the higher prevalence of vp17s in people living with HIV-1 (PLWH) with lymphoma compared with those without lymphoma, suggesting that vp17s may play a key role in lymphomagenesis. Molecular mechanisms involved in vp17 development are still unknown. Here we assessed the efficiency of HIV-1 Reverse Transcriptase (RT) in processing this genomic region and highlighted the existence of hot spots of mutation in Gag, at the end of the matrix protein and close to the matrix-capsid junction. This is possibly due to the presence of inverted repeats and palindromic sequences together with a high content of Adenine in the 322-342 nucleotide portion, which constrain HIV-1 RT to pause on the template. To define the recombinogenic properties of hot spots of mutation in the matrix gene, we developed plasmid vectors expressing Gag and a minimally modified Gag variant, and measured homologous recombination following cell co-nucleofection by next-generation sequencing. Data obtained allowed us to show that a wide range of recombination events occur in concomitance with the identified hot spots of mutation and that imperfect events may account for vp17s generation.
Collapse
Affiliation(s)
- Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Serena Messali
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Matteo Uggeri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Rondina
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Leonardo Sclavi
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Center for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mures, Romania
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Center for Advanced Medical and Pharmaceutical Research (CCAMF), George Emil Palade University of Medicine, Pharmacy, Science and Technology, Târgu Mures, Romania
| |
Collapse
|
3
|
Aryal CM, Pan J. Probing the interactions of the HIV-1 matrix protein-derived polybasic region with lipid bilayers: insights from AFM imaging and force spectroscopy. EUROPEAN BIOPHYSICS JOURNAL : EBJ 2024; 53:57-67. [PMID: 38172352 DOI: 10.1007/s00249-023-01697-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/18/2023] [Accepted: 12/14/2023] [Indexed: 01/05/2024]
Abstract
The human immunodeficiency virus type 1 (HIV-1) matrix protein contains a highly basic region, MA-HBR, crucial for various stages of viral replication. To elucidate the interactions between the polybasic peptide MA-HBR and lipid bilayers, we employed liquid-based atomic force microscopy (AFM) imaging and force spectroscopy on lipid bilayers of differing compositions. In 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC) bilayers, AFM imaging revealed the formation of annulus-shaped protrusions upon exposure to the polybasic peptide, accompanied by distinctive mechanical responses characterized by enhanced bilayer puncture forces. Importantly, our AFM-based force spectroscopy measurements unveiled that MA-HBR induces interleaflet decoupling within the cohesive bilayer organization. This is evidenced by a force discontinuity observed within the bilayer's elastic deformation regime. In POPC/cholesterol bilayers, MA-HBR caused similar yet smaller annular protrusions, demonstrating an intriguing interplay with cholesterol-rich membranes. In contrast, in bilayers containing anionic 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-L-serine (POPS) lipids, MA-HBR induced unique annular protrusions, granular nanoparticles, and nanotubules, showcasing its distinctive effects in anionic lipid-enriched environments. Notably, our force spectroscopy data revealed that anionic POPS lipids weakened interleaflet adhesion within the bilayer, resulting in interleaflet decoupling, which potentially contributes to the specific bilayer perturbations induced by MA-HBR. Collectively, our findings highlight the remarkable variations in how the polybasic peptide, MA-HBR, interacts with lipid bilayers of differing compositions, shedding light on its role in host membrane restructuring during HIV-1 infection.
Collapse
Affiliation(s)
- Chinta M Aryal
- Department of Physics, University of South Florida, Tampa, FL, 33620, USA
- , 2920 Burnet Ave Apt 3, Cincinnati, OH, 45219, USA
| | - Jianjun Pan
- Department of Physics, University of South Florida, Tampa, FL, 33620, USA.
| |
Collapse
|
4
|
HIV-1 mutants expressing B cell clonogenic matrix protein p17 variants are increasing their prevalence worldwide. Proc Natl Acad Sci U S A 2022; 119:e2122050119. [PMID: 35763571 PMCID: PMC9271197 DOI: 10.1073/pnas.2122050119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
In the combined antiretroviral therapy era, lymphomas still represent the most frequent cause of death in HIV-1-infected patients. We expand previous observations dealing with the prevalence of HIV-1 matrix protein p17 variants (vp17s), characterized by peculiar amino acid insertions and endowed of B cell clonogenic activity, in HIV-1 patients with lymphoma as compared with patients without lymphoma. We show that the prevalence of HIV-1 mutants expressing vp17s is increasing worldwide over time. Moreover, we describe a cluster of HIV-1 mutants expressing a B cell clonogenic vp17 and highlight that insertions can be fixed and that viruses displaying clonogenic vp17s are actively spreading. This knowledge advocates for an extensive genomic surveillance program to monitor the evolution of such mutant virions worldwide. AIDS-defining cancers declined after combined antiretroviral therapy (cART) introduction, but lymphomas are still elevated in HIV type 1 (HIV-1)–infected patients. In particular, non-Hodgkin’s lymphomas (NHLs) represent the majority of all AIDS-defining cancers and are the most frequent cause of death in these patients. We have recently demonstrated that amino acid (aa) insertions at the HIV-1 matrix protein p17 COOH-terminal region cause protein destabilization, leading to conformational changes. Misfolded p17 variants (vp17s) strongly impact clonogenic B cell growth properties that may contribute to B cell lymphomagenesis as suggested by the significantly higher frequency of detection of vp17s with COOH-terminal aa insertions in plasma of HIV-1–infected patients with NHL. Here, we expand our previous observations by assessing the prevalence of vp17s in large retrospective cohorts of patients with and without lymphoma. We confirm the significantly higher prevalence of vp17s in lymphoma patients than in HIV-1–infected individuals without lymphoma. Analysis of 3,990 sequences deposited between 1985 and 2017 allowed us to highlight a worldwide increasing prevalence of HIV-1 mutants expressing vp17s over time. Since genomic surveillance uncovered a cluster of HIV-1 expressing a B cell clonogenic vp17 dated from 2011 to 2019, we conclude that aa insertions can be fixed in HIV-1 and that mutant viruses displaying B cell clonogenic vp17s are actively spreading.
Collapse
|
5
|
The relationship of antibodies detected in the Western Blot test with clinical and immunological stages in HIV-infected patients. JOURNAL OF SURGERY AND MEDICINE 2022. [DOI: 10.28982/josam.1005915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
6
|
Abstract
Human immunodeficiency virus type-1(HIV-1)-associated neurocognitive disorder (HAND) remains an important neurological manifestation in HIV-1-infected (HIV+) patients. Furthermore, the HIV-1 matrix protein p17 (p17) detection in the central nervous system (CNS) and its ability to form toxic assemblies in the brain has been recently confirmed. Here we show for the first time using both an in vitro blood-brain barrier (BBB) model and in vivo biodistribution studies in healthy mice that p17 can cross the BBB. There is fast brain uptake with 0.35 ± 0.19% of injected activity per gram of tissue (I.A./g) two minutes after administration, followed by brain accumulation with 0.28 ± 0.09% I.A./g after 1 h. The interaction of p17 with the chemokine receptor 2 (CXCR2) at the surface of brain endothelial cells triggers transcytosis. The present study supports the hypothesis of a direct role of free p17 in neuronal dysfunction in HAND by demonstrating its intrinsic ability to reach the CNS. IMPORTANCE The number of patients affected by HIV-1-associated neurocognitive disorder (HAND) ranges from 30 to 50% of HIV-infected (HIV+) patients. The mechanisms leading to HAND development need to be elucidated, but the role of secreted viral proteins, chemokines, and proinflammatory molecules appears to be clear. In particular, the blood-brain barrier (BBB) represents a route for entry into the central nervous system (CNS) thus playing an important role in HAND. Several findings suggest a key role for the HIV-1 matrix protein p17 (p17) as a microenvironmental factor capable of inducing neurocognitive disorders. Here we show, the ability of the p17 to cross the BBB and to reach the CNS thus playing a crucial role in neuronal dysfunction in HAND.
Collapse
|
7
|
Bacteria-Based Microdevices for the Oral Delivery of Macromolecules. Pharmaceutics 2021; 13:pharmaceutics13101610. [PMID: 34683903 PMCID: PMC8537518 DOI: 10.3390/pharmaceutics13101610] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 12/13/2022] Open
Abstract
The oral delivery of macromolecules is quite challenging due to environmental insults and biological barriers encountered along the gastrointestinal (GI) tract. Benefiting from their living characteristics, diverse bacterial species have been engineered as intelligent platforms to deliver various therapeutics. To tackle difficulties in oral delivery, innovative bacteria-based microdevices have been developed by virtue of advancements in synthetic biology and nanotechnology, with aims to overcome the instability and short half-life of macromolecules in the GI tract. In this review, we summarize the main classes of macromolecules that are produced and delivered through the oral ingestion of bacteria and bacterial derivatives. Furtherly, we discuss the engineering strategies and biomedical applications of these living microdevices in disease diagnosis, bioimaging, and treatment. Finally, we highlight the advantages as well as the limitations of these engineered bacteria used as platforms for the oral delivery of macromolecules and also propose their potential for clinical translation. The results summarized in this review article would contribute to the invention of next-generation bacteria-based systems for the oral delivery of macromolecules.
Collapse
|
8
|
Kammers K, Chen A, Monaco DR, Hudelson SE, Grant-McAuley W, Moore RD, Alter G, Deeks SG, Morrison CS, Eller LA, Blankson JN, Laeyendecker O, Ruczinski I, Eshleman SH, Larman HB. HIV Antibody Profiles in HIV Controllers and Persons With Treatment-Induced Viral Suppression. Front Immunol 2021; 12:740395. [PMID: 34512672 PMCID: PMC8428532 DOI: 10.3389/fimmu.2021.740395] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/10/2021] [Indexed: 11/25/2022] Open
Abstract
Introduction Low HIV viral load is associated with delayed disease progression and reduced HIV transmission. HIV controllers suppress viral load to low levels in the absence of antiretroviral treatment (ART). We used an antibody profiling system, VirScan, to compare antibody reactivity and specificity in HIV controllers, non-controllers with treatment-induced viral suppression, and viremic non-controllers. Methods The VirScan library contains 3,384 phage-displayed peptides spanning the HIV proteome. Antibody reactivity to these peptides was measured in plasma from a Discovery Cohort that included 13 elite controllers, 27 viremic controllers, 12 viremic non-controllers, and 21 non-controllers who were virally suppressed on ART. Antibody reactivity to selected peptides was also assessed in an independent cohort of 29 elite controllers and 37 non-controllers who were virally suppressed on ART (Validation Cohort) and in a longitudinal cohort of non-controllers. Results In the Discovery Cohort, 62 peptides were preferentially targeted in HIV controllers compared to non-controllers who were virally suppressed on ART. These specificities were not significantly different when comparing controllers versus viremic non-controllers. Aggregate reactivity to these peptides was also high in elite controllers from the independent Validation Cohort. The 62 peptides formed seven clusters of homologous epitopes in env, gag, integrase, and vpu. Reactivity to one of these clusters located in gag p17 was inversely correlated with viral load set point in an independent cohort of non-controllers. Conclusions Antibody reactivity was low in non-controllers suppressed on ART, but remained high in viremic controllers despite viral suppression. Antibodies in controllers and viremic non-controllers were directed against epitopes in diverse HIV proteins; higher reactivity against p17 peptides was associated with lower viral load set point. Further studies are needed to determine if these antibodies play a role in regulation of HIV viral load.
Collapse
Affiliation(s)
- Kai Kammers
- Division of Biostatistics and Bioinformatics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Athena Chen
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daniel R. Monaco
- Department of Pathology and the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E. Hudelson
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wendy Grant-McAuley
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Richard D. Moore
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Galit Alter
- Department of Medicine, Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Steven G. Deeks
- Department of Medicine, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Charles S. Morrison
- Behavioral, Epidemiologic and Clinical Sciences, Family Health International (FHI) 360, Durham, NC, United States
| | - Leigh A. Eller
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, United States,Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, United States
| | - Joel N. Blankson
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Oliver Laeyendecker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States,Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Baltimore, MD, United States
| | - Ingo Ruczinski
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Susan H. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: H. Benjamin Larman, ; Susan H. Eshleman,
| | - H. Benjamin Larman
- Department of Pathology and the Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: H. Benjamin Larman, ; Susan H. Eshleman,
| |
Collapse
|
9
|
Isaguliants M, Bayurova E, Avdoshina D, Kondrashova A, Chiodi F, Palefsky JM. Oncogenic Effects of HIV-1 Proteins, Mechanisms Behind. Cancers (Basel) 2021; 13:305. [PMID: 33467638 PMCID: PMC7830613 DOI: 10.3390/cancers13020305] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/28/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
People living with human immunodeficiency virus (HIV-1) are at increased risk of developing cancer, such as Kaposi sarcoma (KS), non-Hodgkin lymphoma (NHL), cervical cancer, and other cancers associated with chronic viral infections. Traditionally, this is linked to HIV-1-induced immune suppression with depletion of CD4+ T-helper cells, exhaustion of lymphopoiesis and lymphocyte dysfunction. However, the long-term successful implementation of antiretroviral therapy (ART) with an early start did not preclude the oncological complications, implying that HIV-1 and its antigens are directly involved in carcinogenesis and may exert their effects on the background of restored immune system even when present at extremely low levels. Experimental data indicate that HIV-1 virions and single viral antigens can enter a wide variety of cells, including epithelial. This review is focused on the effects of five viral proteins: envelope protein gp120, accessory protein negative factor Nef, matrix protein p17, transactivator of transcription Tat and reverse transcriptase RT. Gp120, Nef, p17, Tat, and RT cause oxidative stress, can be released from HIV-1-infected cells and are oncogenic. All five are in a position to affect "innocent" bystander cells, specifically, to cause the propagation of (pre)existing malignant and malignant transformation of normal epithelial cells, giving grounds to the direct carcinogenic effects of HIV-1.
Collapse
Affiliation(s)
- Maria Isaguliants
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden;
- Department of Research, Riga Stradins University, LV-1007 Riga, Latvia
| | - Ekaterina Bayurova
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Darya Avdoshina
- Gamaleya Research Center for Epidemiology and Microbiology, 123098 Moscow, Russia; (E.B.); (D.A.)
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Alla Kondrashova
- M.P. Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819 Moscow, Russia;
| | - Francesca Chiodi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden;
| | - Joel M. Palefsky
- Department of Medicine, University of California, San Francisco, CA 94117, USA;
| |
Collapse
|
10
|
Bugatti A, Paiardi G, Urbinati C, Chiodelli P, Orro A, Uggeri M, Milanesi L, Caruso A, Caccuri F, D'Ursi P, Rusnati M. Heparin and heparan sulfate proteoglycans promote HIV-1 p17 matrix protein oligomerization: computational, biochemical and biological implications. Sci Rep 2019; 9:15768. [PMID: 31673058 PMCID: PMC6823450 DOI: 10.1038/s41598-019-52201-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022] Open
Abstract
p17 matrix protein released by HIV+ cells interacts with leukocytes heparan sulfate proteoglycans (HSPGs), CXCR1 and CXCR2 exerting different cytokine-like activities that contribute to AIDS pathogenesis. Since the bioactive form of several cytokines is represented by dimers/oligomers and oligomerization is promoted by binding to heparin or HSPGs, here we evaluated if heparin/HSPGs also promote p17 oligomerization. Heparin favours p17 dimer, trimer and tetramer assembly, in a time- and biphasic dose-dependent way. Heparin-induced p17 oligomerization is of electrostatic nature, being it prevented by NaCl, by removing negative sulfated groups of heparin and by neutralizing positive lysine residues in the p17 N-terminus. A new computational protocol has been implemented to study heparin chains up to 24-mer accommodating a p17 dimer. Molecular dynamics show that, in the presence of heparin, two p17 molecules undergo conformational modifications creating a continuous “electropositive channel” in which heparin sulfated groups interact with p17 basic amino acids, promoting its dimerization. At the cell surface, HSPGs induce p17 oligomerization, as demonstrated by using B-lymphoblastoid Namalwa cells overexpressing the HSPG Syndecan-1. Also, HSPGs on the surface of BJAB and Raji human B-lymphoblastoid cells are required to p17 to induce ERK1/2 activation, suggesting that HS-induced oligomerization plays a role in p17-induced lymphoid dysregulation during AIDS.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Giulia Paiardi
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Chiara Urbinati
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Paola Chiodelli
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Alessandro Orro
- Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Matteo Uggeri
- Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Luciano Milanesi
- Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Milan, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy
| | - Pasqualina D'Ursi
- Institute for Biomedical Technologies-National Research Council (ITB-CNR), Segrate, Milan, Italy.
| | - Marco Rusnati
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, School of Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
11
|
Liu D, Zeinolabediny Y, Caccuri F, Ferris G, Fang WH, Weston R, Krupinski J, Colombo L, Salmona M, Corpas R, Sarroca S, Sanfeliu C, Caruso A, Guo B, Zeng X, Slevin M. p17 from HIV induces brain endothelial cell angiogenesis through EGFR-1-mediated cell signalling activation. J Transl Med 2019; 99:180-190. [PMID: 30390010 DOI: 10.1038/s41374-018-0147-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/17/2022] Open
Abstract
HIV-associated neurocognitive disorder in HIV patients substantially reduces their quality of life. We previously showed that the HIV matrix protein, p17 could stimulate lymph-angiogenesis in vitro potentially contributing to lymphoma tumour growth and in addition is associated with vascular activation in neuro-degenerating brain tissue; here, therefore, we have investigated the detailed molecular mechanisms of this action. We performed in vitro cell culture, angiogenesis experiments, phospho-protein microarrays and Western blotting to identify cellular signalling induced by p17 within human brain endothelial cells (HbMEC), and inhibitor studies to block p17-induced vascular growth. We also characterised the effects of hippocampal CA1 injection of p17 on epidermal growth factor receptor-1 (EGFR1) expression linked to our murine model of dementia. p17 strongly induced angiogenesis of HbMEC (migration, tube formation and spheroid growth). p17 concomitantly increased phosphorylation of EGFR1 as well as down-stream intermediates ERK1/2, FAK, PLC-γ and PKC-β whilst an inhibitor peptide of EGFR, blocked cell signalling and angiogenesis. Finally, Mice that showed reduced cognitive function and behavioural deficiencies after p17 injection, demonstrated that p17 localised in cortical microvessels and also neurones many of which stained positive for p-EGFR1 by histology/IHC. This work provides strong support that p17 may be involved in initiating and/or perpetuating vascular tissue pathophysiology associated with comorbidity in HIV patients.
Collapse
Affiliation(s)
- Donghui Liu
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.,University of Medicine and Pharmacy, Targu Mures, Romania
| | - Yasmin Zeinolabediny
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.,University of Medicine and Pharmacy, Targu Mures, Romania
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, Section of Microbiology, University of Brescia Medical School, Brescia, Italy
| | - Glenn Ferris
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Wen-Hui Fang
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Ria Weston
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Jerzy Krupinski
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.,Hospital Universitari Mútua de Terrassa, Department of Neurology, Terrassa, Barcelona, Spain
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Ruben Corpas
- Instituto De Investigaciones Biomedicas De Barcelona, CSIC, Barcelona, Spain
| | - Sara Sarroca
- Instituto De Investigaciones Biomedicas De Barcelona, CSIC, Barcelona, Spain
| | - Coral Sanfeliu
- Instituto De Investigaciones Biomedicas De Barcelona, CSIC, Barcelona, Spain
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, Section of Microbiology, University of Brescia Medical School, Brescia, Italy
| | - Baoqiang Guo
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | | | - Mark Slevin
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK. .,University of Medicine and Pharmacy, Targu Mures, Romania. .,Weifang Medical University, Weifang, China.
| |
Collapse
|
12
|
Zhang Z, Zhang F, Bai S, Qiao J, Shen H, Huang F, Gao S, Li S, Gu Y, Xia N. Characterization and epitope mapping of a panel of monoclonal antibodies against HIV‐1 matrix protein. Biotechnol Appl Biochem 2018; 65:807-815. [DOI: 10.1002/bab.1662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 04/09/2018] [Indexed: 11/11/2022]
Affiliation(s)
- Zhiqing Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
| | - Feng Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
| | - Shimeng Bai
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| | - Jiaming Qiao
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
| | - Honglin Shen
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| | - Fang Huang
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| | - Shuangquan Gao
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
| | - Shaowei Li
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| | - Ying Gu
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| | - Ningshao Xia
- State Key Laboratory of Molecular Vaccinology and Molecular DiagnosticsSchool of Public HealthXiamen University Xiamen People's Republic of China
- National Institute of Diagnostics and Vaccine Development in Infectious DiseaseSchool of Life SciencesXiamen University Xiamen People's Republic of China
| |
Collapse
|
13
|
He W, Mazzuca P, Yuan W, Varney K, Bugatti A, Cagnotto A, Giagulli C, Rusnati M, Marsico S, Diomede L, Salmona M, Caruso A, Lu W, Caccuri F. Identification of amino acid residues critical for the B cell growth-promoting activity of HIV-1 matrix protein p17 variants. Biochim Biophys Acta Gen Subj 2018; 1863:13-24. [PMID: 30248376 DOI: 10.1016/j.bbagen.2018.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/18/2018] [Accepted: 09/19/2018] [Indexed: 10/28/2022]
Abstract
BACKGROUND HIV-1 matrix protein p17 variants (vp17s) detected in HIV-1-infected patients with non-Hodgkin's lymphoma (HIV-NHL) display, differently from the wild-type protein (refp17), B cell growth-promoting activity. Biophysical analysis revealed that vp17s are destabilized as compared to refp17, motivating us to explore structure-function relationships. METHODS We used: biophysical techniques (circular dichroism (CD), nuclear magnetic resonance (NMR) and thermal/GuHCL denaturation) to study protein conformation and stability; Surface plasmon resonance (SPR) to study interactions; Western blot to investigate signaling pathways; and Colony Formation and Soft Agar assays to study B cell proliferation and clonogenicity. RESULTS By forcing the formation of a disulfide bridge between Cys residues at positions 57 and 87 we obtained a destabilized p17 capable of promoting B cell proliferation. This finding prompted us to dissect refp17 to identify the functional epitope. A synthetic peptide (F1) spanning from amino acid (aa) 2 to 21 was found to activate Akt and promote B cell proliferation and clonogenicity. Three positively charged aa (Arg15, Lys18 and Arg20) proved critical for sustaining the proliferative activity of both F1 and HIV-NHL-derived vp17s. Lack of any interaction of F1 with the known refp17 receptors suggests an alternate one involved in cell proliferation. CONCLUSIONS The molecular reasons for the proliferative activity of vp17s, compared to refp17, relies on the exposure of a functional epitope capable of activating Akt. GENERAL SIGNIFICANCE Our findings pave the way for identifying the receptor(s) responsible for B cell proliferation and offer new opportunities to identify novel treatment strategies in combating HIV-related NHL.
Collapse
Affiliation(s)
- Wangxiao He
- Center for Translational Medicine, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China.
| | - Pietro Mazzuca
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| | - Weirong Yuan
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA.
| | - Kristen Varney
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA.
| | - Antonella Bugatti
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| | - Alfredo Cagnotto
- IRCCS Istituto Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| | - Cinzia Giagulli
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| | - Marco Rusnati
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy.
| | - Luisa Diomede
- IRCCS Istituto Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| | - Mario Salmona
- IRCCS Istituto Ricerche Farmacologiche "Mario Negri", Milan, Italy.
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| | - Wuyuan Lu
- Center for Translational Medicine, Xi'an Jiaotong University School of Life Science and Technology, Xi'an, China; Institute of Human Virology, University of Maryland School of Medicine, Baltimore, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, USA.
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy.
| |
Collapse
|
14
|
Expression, Purification and Characterization of Hiv-1 Capsid Precursor Protein p41. Protein J 2018; 37:194-202. [PMID: 29508209 DOI: 10.1007/s10930-018-9763-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) has been a global epidemic since 1983; yet, the virology and immunology related to HIV-1 remain elusive. Furthermore, as there is still no effective chemoprophylaxis or vaccine to treat patients with HIV-1, most research focuses on strategies to prevent HIV-1 infection, such as with antiviral drugs, novel therapeutics, or improved diagnostic kits. The HIV-1 Gag precursor protein (p55)-comprising the matrix (MA/p17), capsid (CA/p24), and nucleocapsid (NC/p7) protein domains-is the main structural HIV-1 protein, and is uniquely responsible for virion assembly within the virus life cycle. Recently, the immature and mature capsid structures were solved; however, the precursor protein structure is still unknown. Here, we expressed two subtypes of HIV-1 MA-CA stretch of the Gag protein, referred to as p41, in a bacterial expression system. We characterized the purified p41 protein, and showed its superior antigenicity over that of p24, highlighting the potential influence of the p17 domain on p24 structure. We further showed that p41 has good immunogenicity to induce an antibody response in mice. These results will aid future investigations into the HIV-1 capsid precursor structure, and potentially contribute to improving the design of diagnostic kits.
Collapse
|
15
|
Porchetta A, Ippodrino R, Marini B, Caruso A, Caccuri F, Ricci F. Programmable Nucleic Acid Nanoswitches for the Rapid, Single-Step Detection of Antibodies in Bodily Fluids. J Am Chem Soc 2018; 140:947-953. [PMID: 29313682 DOI: 10.1021/jacs.7b09347] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Antibody detection plays a pivotal role in the diagnosis of pathogens and monitoring the success of vaccine immunization. However, current serology techniques require multiple, time-consuming washing and incubation steps, which limit their applicability in point-of-care (POC) diagnostics and high-throughput assays. We developed here a nucleic acid nanoswitch platform able to instantaneously measure immunoglobulins of type G and E (IgG and IgE) levels directly in blood serum and other bodily fluids. The system couples the advantages of target-binding induced colocalization and nucleic acid conformational-change nanoswitches. Due to the modular nature of the recognition platform, the method can potentially be applied to the detection of any antibody for which an antigen can be conjugated to a nucleic acid strand. In this work we show the sensitive, fast and cost-effective detection of four different antibodies and demonstrate the possible use of this approach for the monitoring of antibody levels in HIV+ patients immunized with AT20 therapeutic vaccine.
Collapse
Affiliation(s)
- Alessandro Porchetta
- Department of Chemistry, University of Rome , Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| | - Rudy Ippodrino
- Ulisse BioMed S.r.l. , Area Science Park, 34149 Trieste, Italy
| | - Bruna Marini
- Ulisse BioMed S.r.l. , Area Science Park, 34149 Trieste, Italy
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, Section of Microbiology, University of Brescia Medical School , Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, Section of Microbiology, University of Brescia Medical School , Piazzale Spedali Civili 1, 25123 Brescia, Italy
| | - Francesco Ricci
- Department of Chemistry, University of Rome , Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy
| |
Collapse
|
16
|
Caccuri F, Giordano F, Barone I, Mazzuca P, Giagulli C, Andò S, Caruso A, Marsico S. HIV-1 matrix protein p17 and its variants promote human triple negative breast cancer cell aggressiveness. Infect Agent Cancer 2017; 12:49. [PMID: 29021819 PMCID: PMC5613317 DOI: 10.1186/s13027-017-0160-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/14/2017] [Indexed: 12/22/2022] Open
Abstract
Background The introduction of cART has changed the morbidity and mortality patterns affecting HIV-infected (HIV+) individuals. The risk of breast cancer in HIV+ patients has now approached the general population risk. However, breast cancer has a more aggressive clinical course and poorer outcome in HIV+ patients than in general population, without correlation with the CD4 or virus particles count. These findings suggest a likely influence of HIV-1 proteins on breast cancer aggressiveness and progression. The HIV-1 matrix protein (p17) is expressed in different tissues and organs of successfully cART-treated patients and promotes migration of different cells. Variants of p17 (vp17s), characterized by mutations and amino acid insertions, differently from the prototype p17 (refp17), also promote B-cell proliferation and transformation. Methods Wound-healing assay, matrigel-based invasion assay, and anchorage-independent proliferation assay were employed to compare the biological activity exerted by refp17 and three different vp17s on the triple-negative human breast cancer cell line MDA-MB 231. Intracellular signaling was investigated by western blot analysis. Results Motility and invasiveness increased in cells treated with both refp17 and vp17s compared to untreated cells. The effects of the viral proteins were mediated by binding to the chemokine receptor CXCR2 and activation of the ERK1/2 signaling pathway. However, vp17s promoted MDA-MB 231 cell growth and proliferation in contrast to refp17-treated or not treated cells. Conclusions In the context of the emerging role of the microenvironment in promoting and supporting cancer cell growth and metastatic spreading, here we provide the first evidence that exogenous p17 may play a crucial role in sustaining breast cancer cell migration and invasiveness, whereas some p17 variants may also be involved in cancer cell growth and proliferation.
Collapse
Affiliation(s)
- Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Francesca Giordano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Pietro Mazzuca
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Cinzia Giagulli
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia , Brescia, Italy
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
17
|
Zeinolabediny Y, Caccuri F, Colombo L, Morelli F, Romeo M, Rossi A, Schiarea S, Ciaramelli C, Airoldi C, Weston R, Donghui L, Krupinski J, Corpas R, García-Lara E, Sarroca S, Sanfeliu C, Slevin M, Caruso A, Salmona M, Diomede L. HIV-1 matrix protein p17 misfolding forms toxic amyloidogenic assemblies that induce neurocognitive disorders. Sci Rep 2017; 7:10313. [PMID: 28871125 PMCID: PMC5583282 DOI: 10.1038/s41598-017-10875-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 08/16/2017] [Indexed: 12/26/2022] Open
Abstract
Human immunodeficiency virus type-1 (HIV-1)-associated neurocognitive disorder (HAND) remains an important neurological manifestation that adversely affects a patient’s quality of life. HIV-1 matrix protein p17 (p17) has been detected in autoptic brain tissue of HAND individuals who presented early with severe AIDS encephalopathy. We hypothesised that the ability of p17 to misfold may result in the generation of toxic assemblies in the brain and may be relevant for HAND pathogenesis. A multidisciplinary integrated approach has been applied to determine the ability of p17 to form soluble amyloidogenic assemblies in vitro. To provide new information into the potential pathogenic role of soluble p17 species in HAND, their toxicological capability was evaluated in vivo. In C. elegans, capable of recognising toxic assemblies of amyloidogenic proteins, p17 induces a specific toxic effect which can be counteracted by tetracyclines, drugs able to hinder the formation of large oligomers and consequently amyloid fibrils. The intrahippocampal injection of p17 in mice reduces their cognitive function and induces behavioral deficiencies. These findings offer a new way of thinking about the possible cause of neurodegeneration in HIV-1-seropositive patients, which engages the ability of p17 to form soluble toxic assemblies.
Collapse
Affiliation(s)
- Yasmin Zeinolabediny
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia, Piazza del Mercato 15, 25121, Brescia, Italy
| | - Laura Colombo
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Federica Morelli
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Margherita Romeo
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Alessandro Rossi
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Silvia Schiarea
- Department of Environmental Health Sciences, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Carlotta Ciaramelli
- Department of Biotechnologies and Biosciences, University of Milano Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Cristina Airoldi
- Department of Biotechnologies and Biosciences, University of Milano Bicocca, Piazza dell'Ateneo Nuovo 1, 20126, Milano, Italy
| | - Ria Weston
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Liu Donghui
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK
| | - Jerzy Krupinski
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.,Hospital Universitari Mútua de Terrassa, Department of Neurology, Terrassa, Barcelona, Spain
| | - Rubén Corpas
- Institut d'Investigaciones Biomèdiques de Barcelona, CSIC and IDIBAPS, Barcelona, Spain
| | - Elisa García-Lara
- Institut d'Investigaciones Biomèdiques de Barcelona, CSIC and IDIBAPS, Barcelona, Spain.,University of Medicine and Pharmacy, Targu Mures, Romania
| | - Sara Sarroca
- Institut d'Investigaciones Biomèdiques de Barcelona, CSIC and IDIBAPS, Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigaciones Biomèdiques de Barcelona, CSIC and IDIBAPS, Barcelona, Spain
| | - Mark Slevin
- School of Healthcare Science, John Dalton Building, Manchester Metropolitan University, Chester Street, Manchester, M1 5GD, UK.,University of Medicine and Pharmacy, Targu Mures, Romania.,Department of Pathology/Medicine, Griffith University, Brisbane, Australia
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia, Piazza del Mercato 15, 25121, Brescia, Italy
| | - Mario Salmona
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy
| | - Luisa Diomede
- Department of Molecular Biochemistry and Pharmacology, IRCCS- Istituto di Ricerche Farmacologiche "Mario Negri", Via G. La Masa 19, 20156, Milano, Italy.
| |
Collapse
|
18
|
Verrier B, Paul S, Terrat C, Bastide L, Ensinas A, Phelip C, Chanut B, Bulens-Grassigny L, Jospin F, Guillon C. Exploiting Natural Cross-reactivity between Human Immunodeficiency Virus (HIV)-1 p17 Protein and Anti-gp41 2F5 Antibody to Induce HIV-1 Neutralizing Responses In Vivo. Front Immunol 2017; 8:770. [PMID: 28713388 PMCID: PMC5491952 DOI: 10.3389/fimmu.2017.00770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 06/16/2017] [Indexed: 11/26/2022] Open
Abstract
Anti-p17 antibodies are able to neutralize human immunodeficiency virus (HIV) entry in a mouse model. In this study, we identified a region of sequence similarity between the epitopes of anti-p17 neutralizing antibodies and anti-gp41 neutralizing 2F5 antibody and verified cross-reactivity between p17 and 2F5 in vitro. The p17 sequence was modified to increase sequence identity between the p17 and 2F5 epitopes, which resulted in enhanced cross-reactivity in vitro. Immunogenicity of wild-type and modified p17 was characterized in a rabbit model. Both wild-type and mutated p17 induced anti-gp41 responses in rabbits; sera from these animals reacted with gp41 from different HIV clades. Moreover, introduction of the 2F5 sequence in p17 resulted in induction of antibodies with partially neutralizing activity. Based upon these data, we suggest that the natural cross-reactivity between HIV-1 p17 protein and 2F5 antibody can be exploited to induce antibodies with neutralizing activity in an animal model.
Collapse
Affiliation(s)
- Bernard Verrier
- Colloidal Vectors and Tissue Transport, UMR5305, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
| | - Stéphane Paul
- Groupe sur l’Immunité des Muqueuses et Agents Pathogènes, EA3064, Faculté de Médecine Jacques Lisfranc, Université de Lyon, Saint-Etienne, France
| | - Céline Terrat
- Colloidal Vectors and Tissue Transport, UMR5305, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
| | - Liza Bastide
- Retroviruses and Structural Biochemistry, UMR5086, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
| | - Agathe Ensinas
- Colloidal Vectors and Tissue Transport, UMR5305, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
- Groupe sur l’Immunité des Muqueuses et Agents Pathogènes, EA3064, Faculté de Médecine Jacques Lisfranc, Université de Lyon, Saint-Etienne, France
| | - Capucine Phelip
- Colloidal Vectors and Tissue Transport, UMR5305, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
| | - Blandine Chanut
- Groupe sur l’Immunité des Muqueuses et Agents Pathogènes, EA3064, Faculté de Médecine Jacques Lisfranc, Université de Lyon, Saint-Etienne, France
| | - Laura Bulens-Grassigny
- Colloidal Vectors and Tissue Transport, UMR5305, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
- Retroviruses and Structural Biochemistry, UMR5086, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
| | - Fabienne Jospin
- Groupe sur l’Immunité des Muqueuses et Agents Pathogènes, EA3064, Faculté de Médecine Jacques Lisfranc, Université de Lyon, Saint-Etienne, France
| | - Christophe Guillon
- Retroviruses and Structural Biochemistry, UMR5086, Institut de Biologie et Chimie des Protéines, Université de Lyon, CNRS, Lyon, France
- *Correspondence: Christophe Guillon,
| |
Collapse
|
19
|
Konagaya Y, Miyakawa R, Sato M, Matsugami A, Watanabe S, Hayashi F, Kigawa T, Nishimura C. Effect of Glu12-His89 Interaction on Dynamic Structures in HIV-1 p17 Matrix Protein Elucidated by NMR. PLoS One 2016; 11:e0167176. [PMID: 27907055 PMCID: PMC5132258 DOI: 10.1371/journal.pone.0167176] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 11/09/2016] [Indexed: 11/18/2022] Open
Abstract
To test the existence of the salt bridge and stability of the HIV-1 p17 matrix protein, an E12A (mutated at helix 1) was established to abolish possible electrostatic interactions. The chemical shift perturbation from the comparison between wild type and E12A suggested the existence of an electrostatic interaction in wild type between E12 and H89 (located in helix 4). Unexpectedly, the studies using urea denaturation indicated that the E12A substitution slightly stabilized the protein. The dynamic structure of E12A was examined under physiological conditions by both amide proton exchange and relaxation studies. The quick exchange method of amide protons revealed that the residues with faster exchange were located at the mutated region, around A12, compared to those of the wild-type protein. In addition, some residues at the region of helix 4, including H89, exhibited faster exchange in the mutant. In contrast, the average values of the kinetic rate constants for amide proton exchange for residues located in all loop regions were slightly lower in E12A than in wild type. Furthermore, the analyses of the order parameter revealed that less flexible structures existed at each loop region in E12A. Interestingly, the structures of the regions including the alpha1-2 loop and helix 5 of E12A exhibited more significant conformational exchanges with the NMR time-scale than those of wild type. Under lower pH conditions, for further destabilization, the helix 1 and alpha2-3 loop in E12A became more fluctuating than at physiological pH. Because the E12A mutant lacks the activities for trimer formation on the basis of the analytical ultra-centrifuge studies on the sedimentation distribution of p17 (Fledderman et al. Biochemistry 49, 9551–9562, 2010), it is possible that the changes in the dynamic structures induced by the absence of the E12-H89 interaction in the p17 matrix protein contributes to a loss of virus assembly.
Collapse
Affiliation(s)
- Yuta Konagaya
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Tokyo, Japan
| | - Rina Miyakawa
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Tokyo, Japan
| | - Masumi Sato
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Tokyo, Japan
| | - Akimasa Matsugami
- Division of Structural and Synthetic Biology, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan
| | - Satoru Watanabe
- Laboratory for Biomolecular Structure and Dynamics, RIKEN Quantitative Biology Center, Yokohama, Kanagawa, Japan
| | - Fumiaki Hayashi
- Division of Structural and Synthetic Biology, RIKEN Center for Life Science Technologies, Yokohama, Kanagawa, Japan
| | - Takanori Kigawa
- Laboratory for Biomolecular Structure and Dynamics, RIKEN Quantitative Biology Center, Yokohama, Kanagawa, Japan
| | - Chiaki Nishimura
- Faculty of Pharmaceutical Sciences, Teikyo Heisei University, Nakano, Tokyo, Japan
- * E-mail:
| |
Collapse
|
20
|
Caccuri F, Iaria ML, Campilongo F, Varney K, Rossi A, Mitola S, Schiarea S, Bugatti A, Mazzuca P, Giagulli C, Fiorentini S, Lu W, Salmona M, Caruso A. Cellular aspartyl proteases promote the unconventional secretion of biologically active HIV-1 matrix protein p17. Sci Rep 2016; 6:38027. [PMID: 27905556 PMCID: PMC5131311 DOI: 10.1038/srep38027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/03/2016] [Indexed: 11/24/2022] Open
Abstract
The human immune deficiency virus type 1 (HIV-1) matrix protein p17 (p17), although devoid of a signal sequence, is released by infected cells and detected in blood and in different organs and tissues even in HIV-1-infected patients undergoing successful combined antiretroviral therapy (cART). Extracellularly, p17 deregulates the function of different cells involved in AIDS pathogenesis. The mechanism of p17 secretion, particularly during HIV-1 latency, still remains to be elucidated. A recent study showed that HIV-1-infected cells can produce Gag without spreading infection in a model of viral latency. Here we show that in Gag-expressing cells, secretion of biologically active p17 takes place at the plasma membrane and occurs following its interaction with phosphatidylinositol-(4,5)-bisphosphate and its subsequent cleavage from the precursor Gag (Pr55Gag) operated by cellular aspartyl proteases. These enzymes operate a more complex Gag polypeptide proteolysis than the HIV-1 protease, thus hypothetically generating slightly truncated or elongated p17s in their C-terminus. A 17 C-terminal residues excised p17 was found to be structurally and functionally identical to the full-length p17 demonstrating that the final C-terminal region of p17 is irrelevant for the protein’s biological activity. These findings offer new opportunities to identify treatment strategies for inhibiting p17 release in the extracellular microenvironment.
Collapse
Affiliation(s)
- Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Maria Luisa Iaria
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Federica Campilongo
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Kristen Varney
- Department of Biochemistry and Molecular Biology, University of Maryland, Baltimore, Maryland, USA
| | - Alessandro Rossi
- IRCCS Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Stefania Mitola
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Silvia Schiarea
- IRCCS Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Antonella Bugatti
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Pietro Mazzuca
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Cinzia Giagulli
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Simona Fiorentini
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| | - Wuyuan Lu
- Institute of Human Virology, University of Maryland, Baltimore, Maryland, USA
| | - Mario Salmona
- IRCCS Istituto di Ricerche Farmacologiche "Mario Negri" Milan, Italy
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia Medical School, Brescia, Italy
| |
Collapse
|
21
|
Henrick BM, Yao XD, Rosenthal KL. HIV-1 Structural Proteins Serve as PAMPs for TLR2 Heterodimers Significantly Increasing Infection and Innate Immune Activation. Front Immunol 2015; 6:426. [PMID: 26347747 PMCID: PMC4541371 DOI: 10.3389/fimmu.2015.00426] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 08/05/2015] [Indexed: 12/12/2022] Open
Abstract
Immune activation is critical to HIV infection and pathogenesis; however, our understanding of HIV innate immune activation remains incomplete. Recently we demonstrated that soluble TLR2 (sTLR2) physically inhibited HIV-induced NFκB activation and inflammation, as well as HIV-1 infection. In light of these findings, we hypothesized that HIV-1 structural proteins may serve as pathogen-associated molecular patterns (PAMPs) for cellular TLR2 heterodimers. These studies made use of primary human T cells and TZMbl cells stably transformed to express TLR2 (TZMbl-2). Our results demonstrated that cells expressing TLR2 showed significantly increased proviral DNA compared to cells lacking TLR2, and mechanistically this may be due to a TLR2-mediated increased CCR5 expression. Importantly, we show that HIV-1 structural proteins, p17, p24, and gp41, act as viral PAMPs signaling through TLR2 and its heterodimers leading to significantly increased immune activation via the NFκB signaling pathway. Using co-immunoprecipitation and a dot blot method, we demonstrated direct protein interactions between these viral PAMPs and TLR2, while only p17 and gp41 bound to TLR1. Specifically, TLR2/1 heterodimer recognized p17 and gp41, while p24 lead to immune activation through TLR2/6. These results were confirmed using TLR2/1 siRNA knock down assays which ablated p17 and gp41-induced cellular activation and through studies of HEK293 cells expressing selected TLRs. Interestingly, our results show in the absence of TLR6, p24 bound to TLR2 and blocked p17 and gp41-induced activation, thus providing a novel mechanism by which HIV-1 can manipulate innate sensing. Taken together, our results identified, for the first time, novel HIV-1 PAMPs that play a role in TLR2-mediated cellular activation and increased proviral DNA. These findings have important implications for our fundamental understanding of HIV-1 immune activation and pathogenesis, as well as HIV-1 vaccine development.
Collapse
Affiliation(s)
- Bethany M Henrick
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, ON , Canada
| | - Xiao-Dan Yao
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, ON , Canada
| | - Kenneth Lee Rosenthal
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University , Hamilton, ON , Canada
| | | |
Collapse
|
22
|
Focà E, Iaria ML, Caccuri F, Fiorentini S, Motta D, Giagulli C, Castelli F, Caruso A. Long-lasting humoral immune response induced in HIV-1-infected patients by a synthetic peptide (AT20) derived from the HIV-1 matrix protein p17 functional epitope. HIV CLINICAL TRIALS 2015; 16:157-62. [PMID: 26057863 DOI: 10.1179/1528433614z.0000000018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE A therapeutic vaccination based on a synthetic peptide (AT20) representative of the HIV-1 matrix protein p17 (p17) functional region, coupled to keyhole limpet hemocyanin (KLH) AT20-KLH was capable of inducing the production of high-avidity antibodies (Abs) toward a previous untargeted p17 hotspot of functional activity in highly active antiretroviral therapy (HAART)-treated HIV-1-infected patients. Since avidity of Abs after immunization and the retention of antigens are important in sustaining the long-lasting production of specific humoral responses, we asked whether AT20-KLH vaccination would result in development of a long-lived immune response. METHODS The long-term duration of Ab response to AT20-KLH has been evaluated in 10 patients previously enrolled for the AT20-KLH vaccination trial at day 898 post-immunization. Ab titer and their avidity was assessed using specifically designed ELISA assays, whereas their neutralizing capacity was estimated in vitro using a 'wound sealing assay'. RESULTS Data obtained show that high titers of specific anti-AT20 Abs were maintained at more than 2 years after the last immunization. Furthermore, these Abs were capable to neutralize exogenous p17, as assessed by ability of sera derived from AT20-KLH-immunized patients to block the ability of p17 to promote cell migration in vitro. CONCLUSION This finding attests for a successful AT20-KLH vaccine molecule formulation and for an effective HAART-dependent Ab persistence.
Collapse
|
23
|
Paopang P, Kasinrerk W, Tayapiwatana C, Seesuriyachan P, Butr-Indr B. Multiparameter optimization method and enhanced production of secreted recombinant single-chain variable fragment against the HIV-1 P17 protein from Escherichia coli by fed-batch fermentation. Prep Biochem Biotechnol 2015; 46:305-12. [PMID: 25831436 DOI: 10.1080/10826068.2015.1031388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The single-chain fragment variable (scFv) was used to produce a completely functional antigen-binding fragment in bacterial systems. The advancements in antibody engineering have simplified the method of producing Fv fragments and made it more efficient and generally relevant. In a previous study, the scFv anti HIV-1 P17 protein was produced by a batch production system, optimized by the sequential simplex optimization method. This study continued that work in order to enhance secreted scFv production by fed-batch cultivation, which supported high volumetric productivity and provided a large amount of scFvs for diagnostic and therapeutic research. The developments in cell culture media and process parameter settings were required to realize the maximum production of cells. This study investigated the combined optimization methods, Plackett-Burman design (PBD) and sequential simplex optimization, with the aim of optimize feed medium. Fed-batch cultivation with an optimal feeding rate was determined. The result demonstrated that a 20-mL/hr feeding rate of the optimized medium can increase cell growth, total protein production, and scFv anti-p17 activity by 4.43, 1.48, and 6.5 times more than batch cultivation, respectively. The combined optimization method demonstrated novel power tools for the optimization strategy of multiparameter experiments.
Collapse
Affiliation(s)
- Porntip Paopang
- a Division of Clinical Microbiology, Department of Medical Technology, Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand
| | - Watchara Kasinrerk
- b Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand.,c Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand
| | - Chatchai Tayapiwatana
- b Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand.,c Medical Biotechnology Unit, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand
| | - Phisit Seesuriyachan
- d Department of Biotechnology, Faculty of Agro-Industry , Chiang Mai University , Chiang Mai , Thailand
| | - Bordin Butr-Indr
- a Division of Clinical Microbiology, Department of Medical Technology, Faculty of Associated Medical Sciences , Chiang Mai University , Chiang Mai , Thailand
| |
Collapse
|
24
|
Detection of HIV-1 matrix protein p17 quasispecies variants in plasma of chronic HIV-1-infected patients by ultra-deep pyrosequencing. J Acquir Immune Defic Syndr 2014; 66:332-9. [PMID: 24732873 DOI: 10.1097/qai.0000000000000164] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The HIV-1 matrix protein p17 (p17MA) is a pleiotropic protein that plays a key role in the HIV-1 life cycle. It has been long believed to have a highly conserved primary amino acid sequence and a well-preserved structural integrity to avoid severe fitness consequences. However, recent data revealed that the carboxy (COOH)-terminus of p17MA possesses high levels of predicted intrinsic disorder, which would subtend to at least partially unfolded status of this region. This finding pointed to the need of investigating p17MA heterogeneity. METHODS The degree of intrapatient variations in the p17MA primary sequence was assessed on plasma viral RNA by using ultra-deep pyrosequencing. RESULTS Data obtained support a complex nature of p17MA quasispecies, with variants present at variable frequency virtually in all patients. Clusters of mutations were scattered along the entire sequence of the viral protein, but they were more frequently detected within the COOH-terminal region of p17MA. Moreover, deletions and insertions also occurred in a restricted area of the COOH-terminal region. CONCLUSIONS On the whole, our data show that the intrapatient level of sequence diversity in the p17MA is much higher than predicted before. Our results pave the way for further studies aimed at unraveling possible correlations between the presence of distinct p17MA variants and peculiar clinical evolutions of HIV-1 disease. The presence of p17MA quasispecies diversity may offer new tools to improve our understanding of the viral adaptation during the natural history of HIV-1 infection.
Collapse
|
25
|
Abstract
OBJECTIVE Monocyte inflammatory processes are fundamental events in AIDS pathogenesis. HIV-1 matrix protein p17, released from infected cells, was found to exert an interleukin (IL)-8 chemokine-like activity on human monocytes, promoting their trafficking and sustaining inflammatory processes, after binding to CXCR1. A haplotype of the CXCR1 gene (CXCR1_300_142) has been associated with slow HIV disease progression. Here, we determine how CXCR1 genetic variations impact on p17 biological activity. DESIGN/METHODS/RESULTS Our results show that Jurkat cells overexpressing CXCR1 or the receptor carrying single polymorphism CXCR1_300 or CXCR1_142 are able to adhere and migrate in response to both IL-8 and p17. On the contrary, Jurkat cells overexpressing CXCR1_300_142 and monocytes of individuals with such CXCR1 polymorphisms lose the capacity to adhere and migrate in response to p17, but not to their physiological ligand IL-8. Surface plasmon resonance (SPR) and multispectral imaging flow cytometry showed that p17 bound with similar affinity to CXCR1 and CXCR1_300_142. Moreover, whereas p17 was able to activate CXCR1, it was incapable of functionally interacting with CXCR1_300_142 by phosphorylating extracellular signal-regulated kinase 1/2, which regulates chemokine-induced cellular responses. Finally, mutagenesis studies showed that, unlike IL-8, p17 does not use Glu-Leu-Arg-like motifs to activate CXCR1. CONCLUSIONS Our results, showing the inability of p17 to activate CXCR1_300_142, a receptor found to be expressed on immune cells of patients with a low progression of HIV disease, point to a crucial role of p17 in AIDS pathogenesis. Our findings herein call for an exploration of the therapeutic potential of blocking the p17/CXCR1 axis in HIV infection.
Collapse
|
26
|
Virus particle release from glycosphingolipid-enriched microdomains is essential for dendritic cell-mediated capture and transfer of HIV-1 and henipavirus. J Virol 2014; 88:8813-25. [PMID: 24872578 DOI: 10.1128/jvi.00992-14] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Human immunodeficiency virus type 1 (HIV-1) exploits dendritic cells (DCs) to promote its transmission to T cells. We recently reported that the capture of HIV-1 by mature dendritic cells (MDCs) is mediated by an interaction between the glycosphingolipid (GSL) GM3 on virus particles and CD169/Siglec-1 on MDCs. Since HIV-1 preferentially buds from GSL-enriched lipid microdomains on the plasma membrane, we hypothesized that the virus assembly and budding site determines the ability of HIV-1 to interact with MDCs. In support of this hypothesis, mutations in the N-terminal basic domain (29/31KE) or deletion of the membrane-targeting domain of the HIV-1 matrix (MA) protein that altered the virus assembly and budding site to CD63(+)/Lamp-1-positive intracellular compartments resulted in lower levels of virion incorporation of GM3 and attenuation of virus capture by MDCs. Furthermore, MDC-mediated capture and transmission of MA mutant viruses to T cells were decreased, suggesting that HIV-1 acquires GSLs via budding from the plasma membrane to access the MDC-dependent trans infection pathway. Interestingly, MDC-mediated capture of Nipah and Hendra virus (recently emerged zoonotic paramyxoviruses) M (matrix) protein-derived virus-like particles that bud from GSL-enriched plasma membrane microdomains was also dependent on interactions between virion-incorporated GSLs and CD169. Moreover, capture and transfer of Nipah virus envelope glycoprotein-pseudotyped lentivirus particles by MDCs were severely attenuated upon depletion of GSLs from virus particles. These results suggest that GSL incorporation into virions is critical for the interaction of diverse enveloped RNA viruses with DCs and that the GSL-CD169 recognition nexus might be a conserved viral mechanism of parasitization of DC functions for systemic virus dissemination. IMPORTANCE Dendritic cells (DCs) can capture HIV-1 particles and transfer captured virus particles to T cells without establishing productive infection in DCs, a mechanism of HIV-1 trans infection. We have recently identified CD169-mediated recognition of GM3, a host-derived glycosphingolipid (GSL) incorporated into the virus particle membrane, as the receptor and ligand for the DC-HIV trans infection pathway. In this study, we have identified the matrix (MA) domain of Gag to be the viral determinant that governs incorporation of GM3 into HIV-1 particles, a previously unappreciated function of the HIV-1 MA. In addition, we demonstrate that the GSL-CD169-dependent trans infection pathway is also utilized as a dissemination mechanism by henipaviruses. GSL incorporation in henipaviruses was also dependent on the viral capsid (M) protein-directed assembly and budding from GSL-enriched lipid microdomains. These findings provide evidence of a conserved mechanism of retrovirus and henipavirus parasitization of cell-to-cell recognition pathways for systemic virus dissemination.
Collapse
|
27
|
Simian immunodeficiency virus and human immunodeficiency virus type 1 matrix proteins specify different capabilities to modulate B cell growth. J Virol 2014; 88:5706-17. [PMID: 24623414 DOI: 10.1128/jvi.03142-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Exogenous HIV-1 matrix protein p17 (p17) deregulates the function of different cells after its N-terminal loop (AT20) binding to the chemokine receptors CXCR1 and CXCR2. One site within AT20 has been recently found to be the major determinant of viral fitness following transmission of simian immunodeficiency virus (SIV) to the human host. Therefore, we sought to determine whether SIV matrix protein (MA) was already capable of interacting with CXCR1 and CXCR2 and mimic p17 biological activities rather than this being a newly acquired function during host adaptation. We show here that SIV MA binds with the same affinity of p17 to CXCR1 and CXCR2 and displays both p17 proangiogenic on human primary endothelial cells and chemotactic activity on human primary monocytes and B cells. However, SIV MA exhibited a higher degree of plasticity than p17 in the C terminus, a region known to play a role in modulating B cell growth. Indeed, in contrast to p17, SIV MA was found to activate the phosphatidylinositol 3-kinase/Akt signaling pathway and strongly promote B cell proliferation and clonogenic activity. Interestingly, we have recently highlighted the existence of a Ugandan HIV-1 strain-derived p17 variant (S75X) with the same B cell growth-promoting activity of SIV MA. Computational modeling allowed us to hypothesize an altered C terminus/core region interaction behind SIV MA and S75X activity. Our findings suggest the appearance of a structural constraint in the p17 C terminus that controls B cell growth, which may help to elucidate the evolutionary trajectory of HIV-1. IMPORTANCE The HIV-1 matrix protein p17 (p17) deregulates the biological activities of different cells after binding to the chemokine receptors CXCR1 and CXCR2. The p17 functional domain responsible for receptors interaction includes an amino acid which is considered the major determinant of SIV replication in humans. Therefore, we sought to determine whether SIV matrix protein (SIV MA) already had the ability to bind to both chemokine receptors rather than being a function newly acquired during host adaptation. We show here that SIV MA binds to CXCR1 and CXCR2 and fully mimics the p17 proangiogenic and chemokine activity. However, it differs from p17 in its ability to signal into B cells and promote B cell growth and clonogenicity. Computational analysis suggests that the accumulation of mutations in the C-terminal region may have led to a further SIV MA adaptation to the human host. This finding in turn sheds light on the evolutionary trajectory of HIV-1.
Collapse
|
28
|
Caccuri F, Rueckert C, Giagulli C, Schulze K, Basta D, Zicari S, Marsico S, Cervi E, Fiorentini S, Slevin M, Guzman CA, Caruso A. HIV-1 matrix protein p17 promotes lymphangiogenesis and activates the endothelin-1/endothelin B receptor axis. Arterioscler Thromb Vasc Biol 2014; 34:846-56. [PMID: 24482377 DOI: 10.1161/atvbaha.113.302478] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE AIDS-related lymphomas are high grade and aggressively metastatic with poor prognosis. Lymphangiogenesis is essential in supporting proliferation and survival of lymphoma, as well as tumor dissemination. Data suggest that aberrant lymphangiogenesis relies on action of HIV-1 proteins rather than on a direct effect of the virus itself. HIV-1 matrix protein p17 was found to accumulate and persist in lymph nodes of patients even under highly active antiretroviral therapy. Because p17 was recently found to exert a potent proangiogenic activity by interacting with chemokine (C-X-C motif) receptors 1 and 2, we tested the prolymphangiogenic activity of the viral protein. APPROACH AND RESULTS Human primary lymph node-derived lymphatic endothelial cells were used to perform capillary-like structure formation, wound healing, spheroids, and Western blot assays after stimulation with or without p17. Here, we show that p17 promotes lymphangiogenesis by binding to chemokine (C-X-C motif) receptor-1 and chemokine (C-X-C motif) receptor-2 expressed on lymph node-derived lymphatic endothelial cells and activating the Akt/extracellular signal-regulated kinase signaling pathway. In particular, it was found to induce capillary-like structure formation, sprout formation from spheroids, and increase lymph node-derived lymphatic endothelial cells motility. The p17 lymphangiogenic activity was, in part, sustained by activation of the endothelin-1/endothelin receptor B axis. A Matrigel plug assay showed that p17 was able to promote the outgrowth of lymphatic vessels in vivo, demonstrating that p17 directly regulates lymphatic vessel formation. CONCLUSIONS Our results suggest that p17 may generate a prolymphangiogenic microenvironment and plays a role in predisposing the lymph node to lymphoma growth and metastasis. This finding offers new opportunities to identify treatment strategies in combating AIDS-related lymphomas.
Collapse
Affiliation(s)
- Francesca Caccuri
- From the Microbiology Section, Department of Molecular and Translational Medicine (F.C., C.G., D.B., S.Z., S.F., A.C.) and Section of Vascular Surgery, Department of Medical and Surgical Sciences (E.C.), University of Brescia, Brescia, Italy; Animal Models and Retroviral Vaccine Section, National Cancer Institute, National Institutes of Health, Bethesda, MD (F.C.); Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Braunschweig, Germany (C.R., K.S., C.A.G.); Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy (S.M.); and School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom (M.S.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Iaria ML, Fiorentini S, Focà E, Zicari S, Giagulli C, Caccuri F, Francisci D, Di Perri G, Castelli F, Baldelli F, Caruso A. Synthetic HIV-1 matrix protein p17-based AT20-KLH therapeutic immunization in HIV-1-infected patients receiving antiretroviral treatment: A phase I safety and immunogenicity study. Vaccine 2014; 32:1072-8. [PMID: 24456624 DOI: 10.1016/j.vaccine.2013.12.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Therapeutic vaccination is a promising novel approach to treat HIV-1 infected people by boosting or redirecting immune system to neutralize critical HIV-1 antigens whose biological effects are relevant in the context of viral pathogenesis. With the aim to induce neutralizing antibodies to the matrix protein p17 we have developed a peptide-based immunogen (AT20-KLH) and evaluated its safety and immunogenicity. METHODOLOGY Twenty four asymptomatic HAART-treated HIV-1+ patients were enrolled in a phase I clinical study and were randomized to three groups: 2 groups were treated with five IM injection (Arm A: 25μg/inoculation; Arm B: 100μg/inoculation) at day (D) D0, D28, D56, D84 and D112; the control group (Arm C) were not injected. Safety was assessed by monitoring local and systemic adverse events (AEs), recorded till D168. Evaluation of immunogenicity was by titering antibodies at D0, D35, D56, D63, D84, D91, D112, D140 and D168 using ELISA. RESULTS In all, 105 local and systemic AEs were reported across the three groups. Most were mild and resolved without sequelae. Also the few unsolicited events, deemed unrelated to the study vaccines, caused no problems. No significant changes in the routine laboratory parameters, CD4 T-cell count or HIV-1 viremia were found. At the time of enrollment 23 out of 24 patients had no anti-AT20 antibodies, whereas 11 exhibited anti-p17 antibodies. Irrespective of the presence of preimmunization antibodies, all subjects developed high titers of anti-AT20 antibodies (GM 9775) in response to both AT20-KLH doses. These antibodies were also capable of recognizing AT20 within the p17 framework. CONCLUSIONS The AT20 peptide-based approach has allowed to redirect HAART-treated patients' humoral responses toward a previously untargeted hotspot of functional activity. Overall, the tested AT20-KLH doses were safe and well tolerated, supporting further exploration of AT20-KLH as an HIV-1 therapeutic vaccine candidate.
Collapse
Affiliation(s)
- Maria Luisa Iaria
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Simona Fiorentini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Emanuele Focà
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Sonia Zicari
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cinzia Giagulli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Francesca Caccuri
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Daniela Francisci
- Section of Infectious Diseases, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, 06132 Perugia, Italy
| | | | - Francesco Castelli
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Franco Baldelli
- Section of Infectious Diseases, Department of Experimental Medicine and Biochemical Sciences, University of Perugia, 06132 Perugia, Italy
| | - Arnaldo Caruso
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
30
|
Flexible and rigid structures in HIV-1 p17 matrix protein monitored by relaxation and amide proton exchange with NMR. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1844:520-6. [PMID: 24373876 DOI: 10.1016/j.bbapap.2013.12.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 12/06/2013] [Accepted: 12/18/2013] [Indexed: 11/22/2022]
Abstract
The HIV-1 p17 matrix protein is a multifunctional protein that interacts with other molecules including proteins and membranes. The dynamic structure between its folded and partially unfolded states can be critical for the recognition of interacting molecules. One of the most important roles of the p17 matrix protein is its localization to the plasma membrane with the Gag polyprotein. The myristyl group attached to the N-terminus on the p17 matrix protein functions as an anchor for binding to the plasma membrane. Biochemical studies revealed that two regions are important for its function: D14-L31 and V84-V88. Here, the dynamic structures of the p17 matrix protein were studied using NMR for relaxation and amide proton exchange experiments at the physiological pH of 7.0. The results revealed that the α12-loop, which includes the 14-31 region, was relatively flexible, and that helix 4, including the 84-88 region, was the most protected helix in this protein. However, the residues in the α34-loop near helix 4 had a low order parameter and high exchange rate of amide protons, indicating high flexibility. This region is probably flexible because this loop functions as a hinge for optimizing the interactions between helices 3 and 4. The C-terminal long region of K113-Y132 adopted a disordered structure. Furthermore, the C-terminal helix 5 appeared to be slightly destabilized due to the flexible C-terminal tail based on the order parameters. Thus, the dynamic structure of the p17 matrix protein may be related to its multiple functions.
Collapse
|
31
|
Bugatti A, Giagulli C, Urbinati C, Caccuri F, Chiodelli P, Oreste P, Fiorentini S, Orro A, Milanesi L, D'Ursi P, Caruso A, Rusnati M. Molecular interaction studies of HIV-1 matrix protein p17 and heparin: identification of the heparin-binding motif of p17 as a target for the development of multitarget antagonists. J Biol Chem 2012; 288:1150-61. [PMID: 23166320 DOI: 10.1074/jbc.m112.400077] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Once released by HIV(+) cells, p17 binds heparan sulfate proteoglycans (HSPGs) and CXCR1 on leukocytes causing their dysfunction. By exploiting an approach integrating computational modeling, site-directed mutagenesis of p17, chemical desulfation of heparin, and surface plasmon resonance, we characterized the interaction of p17 with heparin, a HSPG structural analog, and CXCR1. p17 binds to heparin with an affinity (K(d) = 190 nm) that is similar to those of other heparin-binding viral proteins. Two stretches of basic amino acids (basic motifs) are present in p17 N and C termini. Neutralization (Arg→Ala substitution) of the N-terminal, but not of the C-terminal basic motif, causes the loss of p17 heparin-binding capacity. The N-terminal heparin-binding motif of p17 partially overlaps the CXCR1-binding domain. Accordingly, its neutralization prevents also p17 binding to the chemochine receptor. Competition experiments demonstrated that free heparin and heparan sulfate (HS), but not selectively 2-O-, 6-O-, and N-O desulfated heparins, prevent p17 binding to substrate-immobilized heparin, indicating that the sulfate groups of the glycosaminoglycan mediate p17 interaction. Evaluation of the p17 antagonist activity of a panel of biotechnological heparins derived by chemical sulfation of the Escherichia coli K5 polysaccharide revealed that the highly N,O-sulfated derivative prevents the binding of p17 to both heparin and CXCR1, thus inhibiting p17-driven chemotactic migration of human monocytes with an efficiency that is higher than those of heparin and HS. Here, we characterized at a molecular level the interaction of p17 with its cellular receptors, laying the basis for the development of heparin-mimicking p17 antagonists.
Collapse
Affiliation(s)
- Antonella Bugatti
- Section of Experimental Oncology and Immunology, School of Medicine, University of Brescia, Brescia 25123, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
HIV-1 matrix protein p17 promotes angiogenesis via chemokine receptors CXCR1 and CXCR2. Proc Natl Acad Sci U S A 2012; 109:14580-5. [PMID: 22904195 DOI: 10.1073/pnas.1206605109] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Vascular diseases supported by aberrant angiogenesis have increased incidence in HIV-1-infected patients. Several data suggest that endothelium dysfunction relies on action of HIV-1 proteins rather than on a direct effect of the virus itself. The HIV-1 matrix protein p17 is known to deregulate the biological activity of different immune cells. Recently, p17 was found to mimic IL-8 chemokine activity by binding to the IL-8 receptor CXCR1. Here we show that p17 binds with high affinity to CXCR2, a CXCR1-related receptor, and promotes the formation of capillary-like structures on human endothelial cells (ECs) by interacting with both CXCR1 and CXCR2 expressed on the EC surface. ERK signaling via Akt was defined as the pathway responsible for p17-induced tube formation. Ex vivo and in vivo experimental models confirmed the provasculogenic activity of p17, which was comparable to that induced by VEGF-A. The hypothesis of a major role for p17 in HIV-1-induced aberrant angiogenesis is enforced by the finding that p17 is detected, as a single protein, in blood vessels of HIV-1-patients and in particular in the nucleus of ECs. Localization of p17 in the nucleus of ECs was evidenced also in in vitro experiments, suggesting the internalization of exogenous p17 in ECs by mechanisms of receptor-mediated endocytosis. Recognizing p17 interaction with CXCR1 and CXCR2 as the key event in sustaining EC aberrant angiogenesis could help us to identify new treatment strategies in combating AIDS-related vascular diseases.
Collapse
|
33
|
Renga B, Francisci D, D'Amore C, Schiaroli E, Mencarelli A, Cipriani S, Baldelli F, Fiorucci S. The HIV matrix protein p17 subverts nuclear receptors expression and induces a STAT1-dependent proinflammatory phenotype in monocytes. PLoS One 2012; 7:e35924. [PMID: 22558273 PMCID: PMC3340403 DOI: 10.1371/journal.pone.0035924] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 03/23/2012] [Indexed: 01/22/2023] Open
Abstract
Background Long-term remission of HIV-1 disease can be readily achieved by combinations of highly effective antiretroviral therapy (HAART). However, a residual persistent immune activation caused by circulating non infectious particles or viral proteins is observed under HAART and might contribute to an higher risk of non-AIDS pathologies and death in HIV infected persons. A sustained immune activation supports lipid dysmetabolism and increased risk for development of accelerated atehrosclerosis and ischemic complication in virologically suppressed HIV-infected persons receiving HAART. Aim While several HIV proteins have been identified and characterized for their ability to maintain immune activation, the role of HIV-p17, a matrix protein involved in the viral replication, is still undefined. Results Here, we report that exposure of macrophages to recombinant human p17 induces the expression of proinflammatory and proatherogenic genes (MCP-1, ICAM-1, CD40, CD86 and CD36) while downregulating the expression of nuclear receptors (FXR and PPARγ) that counter-regulate the proinflammatory response and modulate lipid metabolism in these cells. Exposure of macrophage cell lines to p17 activates a signaling pathway mediated by Rack-1/Jak-1/STAT-1 and causes a promoter-dependent regulation of STAT-1 target genes. These effects are abrogated by sera obtained from HIV-infected persons vaccinated with a p17 peptide. Ligands for FXR and PPARγ counteract the effects of p17. Conclusions The results of this study show that HIV p17 highjacks a Rack-1/Jak-1/STAT-1 pathway in macrophages, and that the activation of this pathway leads to a simultaneous dysregulation of immune and metabolic functions. The binding of STAT-1 to specific responsive elements in the promoter of PPARγ and FXR and MCP-1 shifts macrophages toward a pro-atherogenetic phenotype characterized by high levels of expression of the scavenger receptor CD36. The present work identifies p17 as a novel target in HIV therapy and grounds the development of anti-p17 small molecules or vaccines.
Collapse
Affiliation(s)
- Barbara Renga
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Dipartimento di Medicina Clinica e Scienze Biochimiche, University of Perugia, Perugia, Italy
| | - Claudio D'Amore
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Dipartimento di Medicina Clinica e Scienze Biochimiche, University of Perugia, Perugia, Italy
| | - Andrea Mencarelli
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Perugia, Italy
| | - Sabrina Cipriani
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Perugia, Italy
| | - Franco Baldelli
- Dipartimento di Medicina Clinica e Scienze Biochimiche, University of Perugia, Perugia, Italy
| | - Stefano Fiorucci
- Dipartimento di Medicina Clinica e Sperimentale, University of Perugia, Perugia, Italy
- * E-mail:
| |
Collapse
|
34
|
Dahiya S, Nonnemacher MR, Wigdahl B. Deployment of the human immunodeficiency virus type 1 protein arsenal: combating the host to enhance viral transcription and providing targets for therapeutic development. J Gen Virol 2012; 93:1151-1172. [PMID: 22422068 DOI: 10.1099/vir.0.041186-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Despite the success of highly active antiretroviral therapy in combating human immunodeficiency virus type 1 (HIV-1) infection, the virus still persists in viral reservoirs, often in a state of transcriptional silence. This review focuses on the HIV-1 protein and regulatory machinery and how expanding knowledge of the function of individual HIV-1-coded proteins has provided valuable insights into understanding HIV transcriptional regulation in selected susceptible cell types. Historically, Tat has been the most studied primary transactivator protein, but emerging knowledge of HIV-1 transcriptional regulation in cells of the monocyte-macrophage lineage has more recently established that a number of the HIV-1 accessory proteins like Vpr may directly or indirectly regulate the transcriptional process. The viral proteins Nef and matrix play important roles in modulating the cellular activation pathways to facilitate viral replication. These observations highlight the cross talk between the HIV-1 transcriptional machinery and cellular activation pathways. The review also discusses the proposed transcriptional regulation mechanisms that intersect with the pathways regulated by microRNAs and how development of the knowledge of chromatin biology has enhanced our understanding of key protein-protein and protein-DNA interactions that form the HIV-1 transcriptome. Finally, we discuss the potential pharmacological approaches to target viral persistence and enhance effective transcription to purge the virus in cellular reservoirs, especially within the central nervous system, and the novel therapeutics that are currently in various stages of development to achieve a much superior prognosis for the HIV-1-infected population.
Collapse
Affiliation(s)
- Satinder Dahiya
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Michael R Nonnemacher
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA 19129, USA
| |
Collapse
|
35
|
HIV-1 matrix protein p17 binds to the IL-8 receptor CXCR1 and shows IL-8–like chemokine activity on monocytes through Rho/ROCK activation. Blood 2012; 119:2274-83. [DOI: 10.1182/blood-2011-06-364083] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
AbstractExogenous HIV-1 matrix protein p17 was found to deregulate biologic activities of many different immune cells that are directly or indirectly involved in AIDS pathogenesis after binding to unknown cellular receptor(s). In particular, p17 was found to induce a functional program in monocytes related to activation and inflammation. In the present study, we demonstrate that CXCR1 is the receptor molecule responsible for p17 chemokine–like activity on monocytes. After CXCR1 binding, p17 was capable of triggering rapid adhesion and chemotaxis of monocytes through a pathway that involved Rho/ROCK. Moreover, CXCR1-silenced primary monocytes lost responsiveness to p17 chemoattraction, whereas CXCR1-transfected Jurkat cells acquired responsiveness. Surface plasmon resonance studies confirmed the capacity of p17 to bind CXCR1 and showed that the p17/CXCR1 interaction occurred with a low affinity compared with that measured for IL-8, the physiologic CXCR1 ligand. In all of its activities, p17 mimicked IL-8, the natural high-affinity ligand of CXCR1. Recent studies have highlighted the role of IL-8 and CXCR1 in HIV-1 replication and AIDS pathogenesis. Our findings herein call for an exploration of the therapeutic potential of blocking the p17/IL-8/CXCR1 axis in HIV-1 infection.
Collapse
|
36
|
Giagulli C, Marsico S, Magiera AK, Bruno R, Caccuri F, Barone I, Fiorentini S, Andò S, Caruso A. Opposite effects of HIV-1 p17 variants on PTEN activation and cell growth in B cells. PLoS One 2011; 6:e17831. [PMID: 21423810 PMCID: PMC3056727 DOI: 10.1371/journal.pone.0017831] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Accepted: 02/15/2011] [Indexed: 11/18/2022] Open
Abstract
The HIV-1 matrix protein p17 is a structural protein that can act in the extracellular environment to deregulate several functions of immune cells, through the interaction of its NH(2)-terminal region with a cellular surface receptor (p17R). The intracellular events triggered by p17/p17R interaction have been not completely characterized yet. In this study we analyze the signal transduction pathways induced by p17/p17R interaction and show that in Raji cells, a human B cell line stably expressing p17R on its surface, p17 induces a transient activation of the transcriptional factor AP-1. Moreover, it was found to upregulate pERK1/2 and downregulate pAkt, which are the major intracellular signalling components involved in AP-1 activation. These effects are mediated by the COOH-terminal region of p17, which displays the capability of keeping PTEN, a phosphatase that regulates the PI3K/Akt pathway, in an active state through the serine/threonine (Ser/Thr) kinase ROCK. Indeed, the COOH-terminal truncated form of p17 (p17Δ36) induced activation of the PI3K/Akt pathway by maintaining PTEN in an inactive phosphorylated form. Interestingly, we show that among different p17s, a variant derived from a Ugandan HIV-1 strain, named S75X, triggers an activation of PI3K/Akt signalling pathway, and leads to an increased B cell proliferation and malignant transformation. In summary, this study shows the role of the COOH-terminal region in modulating the p17 signalling pathways so highlighting the complexity of p17 binding to and signalling through its receptor(s). Moreover, it provides the first evidence on the presence of a p17 natural variant mimicking the p17Δ36-induced signalling in B cells and displaying the capacity of promoting B cell growth and tumorigenesis.
Collapse
Affiliation(s)
- Cinzia Giagulli
- Department of Experimental and Applied Medicine, University of Brescia, Brescia, Italy
| | - Stefania Marsico
- Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy
| | | | - Rosalinda Bruno
- Department of Pharmaco-Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy
| | - Francesca Caccuri
- Department of Experimental and Applied Medicine, University of Brescia, Brescia, Italy
| | - Ines Barone
- Department of Cell Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy
| | - Simona Fiorentini
- Department of Experimental and Applied Medicine, University of Brescia, Brescia, Italy
| | - Sebastiano Andò
- Department of Cell Biology, University of Calabria, Arcavacata di Rende (Cosenza), Italy
| | - Arnaldo Caruso
- Department of Experimental and Applied Medicine, University of Brescia, Brescia, Italy
- * E-mail:
| |
Collapse
|